Acessibilidade / Reportar erro

Behavioral or neuropsychiatric symptoms of Alzheimer's disease: from psychopathology to pharmacological management

Sintomas comportamentais ou neuropsiquiátricos da doença de Alzheimer: psicopatologia e abordagem terapêutica

Abstract

Neuropsychiatric or behavioral symptoms of dementia encompass a series of disorders, such as anxiety, depression, apathy, psychosis, and agitation, all commonly present in individuals living with dementia. While they are not required for the diagnosis of Alzheimer's disease (AD), they are ubiquitously present in all stages of the disease, contributing to negative clinical outcomes, including cognitive decline, functional disability, and caregiver burden. Neuropsychiatric symptoms have been conceptualized not only as risk factors but as clinical markers of decline along the AD spectrum. The concept of “mild behavioral impairment”, the behavioral correlate of mild cognitive impairment, has been proposed within this framework. The first steps in the management of behavioral symptoms in AD involve defining the target and investigating potential causes and/or aggravating factors. Once these factors are addressed, non-pharmacological approaches are preferred as first-line interventions. Following the optimization of anticholinesterase treatments, specific pharmacological approaches (e.g., antidepressants, antipsychotics) can be considered weighing potential side effects.

Keywords
Alzheimer Disease; Neuropsychiatry; Apathy; Depression; Anxiety; Psychomotor Agitation; Psychotic Disorders; Sleep

Resumo

Sintomas neuropsiquiátricos ou comportamentais de demência envolvem uma série de condições, como ansiedade, depressão, apatia, psicose e agitação, frequentemente observadas em indivíduos com demência. Embora esses sintomas não sejam necessários para o diagnóstico da doença de Alzheimer, estão presentes em todas as fases ou estágios da doença, contribuindo negativamente para o declínio cognitivo, comprometimento funcional e sobrecarga do cuidador. Os sintomas neuropsiquiátricos têm sido conceituados não apenas como fatores de risco, mas também como marcadores clínicos de progressão da doença de Alzheimer. O construto “comprometimento comportamental leve”, correlato comportamental do comprometimento cognitive leve, tem sido proposto nesse contexto. Os primeiros passos na abordagem dos sintomas comportamentais da doença de Alzheimer envolvem definir os alvos-terapêuticos e investigar potenciais causas ou fatores agravantes. Após intervir nesses fatores, abordagens não farmacológicas constituem a primeira linha de intervenção. Depois da otimização do tratamento anticolinesterásico, terapias farmacológicas específicas (por exemplo, antidepressivos, antipsicóticos) podem ser consideradas, levando-se em conta potencias efeitos colaterais.

Palavras-chave
Doença de Alzheimer; Neuropsiquiatria; Apatia; Depressão; Ansiedade; Agitação Psicomotora; Transtornos Psicóticos; Sono

INTRODUCTION

Alzheimer's disease (AD) is the leading cause of dementia, a clinical syndrome characterized by a decline in cognitive functions with functional repercussions.11 Scheltens P, De Strooper B, Kivipelto M, et al. Alzheimer’s disease. Lancet 2021;397(10284):1577-1590 Behavioral symptoms are not required for diagnosis, but they are frequently recognized in AD, especially in its moderate to severe stages.22 Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of Early Alzheimer’s Disease: Clinical Practice in 2021. J Prev Alzheimers Dis 2021;8(03):371-386 Actually, they may occur in all stages of AD, including preclinical and prodromal stages, and virtually all patients will develop at least one behavioral disorder during the course of the disease.33 Steinberg M, Shao H, Zandi P, et al; Cache County Investigators. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry 2008;23(02):170-177 Cross-sectional studies have shown variable prevalence numbers, with rates varying according to sample characteristics, measurement tools, and study setting, with higher rates observed in institutionalized patients.44 Zhao QF, Tan L, Wang HF, et al. The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: Systematic review and meta-analysis. J Affect Disord 2016;190:264-271 The pathophysiological mechanisms underlying AD presentation overlap with those associated with neuropsychiatric syndromes, and the relationship between cognitive and behavioral symptoms is thought to be bidirectional (Figure 1).

Figure 1
AD-related pathophysiological mechanisms potentially link cognitive impairment and behavioral symptoms. Proteinopathy (abnormal accumulation of amyloid-β and tau protein), neuroinflammation, and oxidative stress contribute to neuronal dysfunction and, ultimately, neuronal death in AD. The consequent neurotransmitter imbalance and neural circuit dysfunction result in cognitive impairment and behavioral symptoms. Cognitive and behavioral symptoms tend to aggravate each other.

Behavioral symptoms vary not only according to the stage of neurodegeneration but also the underlying disease.55 Cummings J. The Neuropsychiatric Inventory: Development and Applications. J Geriatr Psychiatry Neurol 2020;33(02):73-84,66 Cummings J. The Role of Neuropsychiatric Symptoms in Research Diagnostic Criteria for Neurodegenerative Diseases. Am J Geriatr Psychiatry 2021;29(04):375-383 For instance, psychosis can affect almost three-quarters of patients with dementia with Lewy bodies, while it is less frequent in AD. Behavioral problems are core features of non-AD types of dementia and usually guide their diagnostic process.55 Cummings J. The Neuropsychiatric Inventory: Development and Applications. J Geriatr Psychiatry Neurol 2020;33(02):73-84,66 Cummings J. The Role of Neuropsychiatric Symptoms in Research Diagnostic Criteria for Neurodegenerative Diseases. Am J Geriatr Psychiatry 2021;29(04):375-383 Identifying vivid visual hallucinations strongly suggests Lewy body disease, while inappropriate social behavior may indicate a behavioral variant of frontotemporal dementia.

The literature usually refers to behavioral symptoms as behavioral and psychological symptoms of dementia (BPSD) or neuropsychiatric symptoms of dementia (NPS).66 Cummings J. The Role of Neuropsychiatric Symptoms in Research Diagnostic Criteria for Neurodegenerative Diseases. Am J Geriatr Psychiatry 2021;29(04):375-383,77 Cerejeira J, Lagarto L, Mukaetova-Ladinska EB. Behavioral and psychological symptoms of dementia. Front Neurol 2012;3:73 These are ‘umbrella’ terms that encompass a series of symptoms, such as agitation, anxiety, apathy, depression, delusion, and hallucination, among others.

Auguste D., the patient originally described by Alois Alzheimer, had more pronounced behavioral problems (related to delusions and hallucinations) than cognitive ones.88 Benjamin S, MacGillivray L, Schildkrout B, Cohen-Oram A, Lauterbach MD, Levin LL. Six Landmark Case Reports Essential for Neuropsychiatric Literacy. J Neuropsychiatry Clin Neurosci 2018;30(04):279-290 In many cases, behavioral symptoms can indeed be more troublesome for patients and caregivers than impaired cognition and the related functional decline. Moreover, behavioral symptoms negatively influence AD-related outcomes, including cognitive decline, functional disability, caregiver burden, costs, medical service utilization, and risk of institutionalization and death.77 Cerejeira J, Lagarto L, Mukaetova-Ladinska EB. Behavioral and psychological symptoms of dementia. Front Neurol 2012;3:73,99 Feast A, Moniz-Cook E, Stoner C, Charlesworth G, Orrell M. A systematic review of the relationship between behavioral and psychological symptoms (BPSD) and caregiver well-being. Int Psychogeriatr 2016;28(11):1761-1774,1010 Sabates J, Chiu WH, Loi S, et al. The Associations Between Neuropsychiatric Symptoms and Cognition in People with Dementia: A Systematic Review and Meta-Analysis. Neuropsychol Rev 2023

Despite their clinical relevance, their management remains challenging, and until very recently, there was no single FDA-approved pharmacological therapy for behavioral symptoms in dementia. The present narrative review will provide an updated perspective of the field, discussing current trends in the classification and treatment of different behaviors along with emerging topics, such as mild behavioral impairment.

MILD BEHAVIORAL IMPAIRMENT

While cognitive symptoms remain the core clinical feature of AD, behavioral symptoms are very common. Behavioral or neuropsychiatric symptoms can occur at any disease stage, and they often manifest very early in the AD continuum, i.e., since its prodromal stages. Their importance was emphasized by the 2011 National Institute on Aging–Alzheimer's Association (NIA-AA) revised guidelines for dementia diagnosis, wherein the 1984 core criteria were modified to explicitly mention “changes in personality, behavior, or comportment—symptoms include uncharacteristic mood fluctuations such as agitation, impaired motivation, initiative, apathy, loss of drive, social withdrawal, decreased interest in previous activities, loss of empathy, compulsive or obsessive behaviors, socially unacceptable behaviors1111 McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011;7(03):263-269.

Behavioral syndromes are also frequently seen as risk factors for AD, increasing the incidence of dementia in both individuals with mild cognitive impairment (MCI) and cognitively normal adults.1212 Geda YE, Roberts RO, Mielke MM, et al. Baseline neuropsychiatric symptoms and the risk of incident mild cognitive impairment: a population-based study. Am J Psychiatry 2014;171(05):572-581,1313 Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202 As acknowledged in the aforementioned criteria, behavioral symptoms have been conceptualized not only as risk factors but as clinical markers of cognitive and functional decline along the AD spectrum. Assuming that neurodegeneration can manifest as behavioral or neuropsychiatric symptoms even before any clinically defined cognitive impairment, Dr. Taragano et al. proposed the mild behavioral impairment (MBI) construct.1414 Taragano FE, Allegri RF, Lyketsos C. Mild behavioral impairment: A prodromal stage of dementia. Dement Neuropsychol 2008;2(04):256-2601616 Taragano FE, Allegri RF, Heisecke SL, et al. Risk of Conversion to Dementia in a Mild Behavioral Impairment Group Compared to a Psychiatric Group and to a Mild Cognitive Impairment Group. J Alzheimers Dis 2018;62(01):227-238 Recently, the International Society to Advance Alzheimer's Research and Treatment (ISTAART) defined MBI as a broad concept that describes a syndrome comprising sustained and impactful late-life neuropsychiatric symptoms (emerging after 50 years of age) that represent a clear change from an individual's baseline and can be early manifestations of neurodegenerative disease,1313 Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202 occurring in cognitively unimpaired individuals or in those with MCI. In other words, MBI can be understood as the behavioral correlate of MCI in the spectrum of neurodegenerative diseases.

MBI comprises five domains: motivation, affective regulation, impulse control, social cognition, and perception/thought content.1313 Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202 For MBI, behavioral symptoms can be of any severity, should precede or occur concurrently with cognitive impairment, and persist for at least six months. By definition, symptoms occurring after the onset of dementia cannot be MBI.1313 Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202,1717 Ismail Z, Agüera-Ortiz L, Brodaty H, et al; NPS Professional Interest Area of the International Society of to Advance Alzheimer’s Research and Treatment (NPS-PIA of ISTAART) The Mild Behavioral Impairment Checklist (MBI-C): A Rating Scale for Neuropsychiatric Symptoms in Pre-Dementia Populations. J Alzheimers Dis 2017;56(03):929-938 The NPS Professional Interest Area of ISTAART developed a rating scale to operationalize MBI. The MBI checklist (MBI-C) is a 34-item instrument that describes and measures changes in a selected list of behavioral symptoms that reflect the five MBI domains of decreased motivation, emotional dysregulation, impulse dyscontrol, social inappropriateness, and abnormal perception or thought content. The MBI-C is a tool intended for use in clinical and research settings to detect and measure changes in NPS over time.1717 Ismail Z, Agüera-Ortiz L, Brodaty H, et al; NPS Professional Interest Area of the International Society of to Advance Alzheimer’s Research and Treatment (NPS-PIA of ISTAART) The Mild Behavioral Impairment Checklist (MBI-C): A Rating Scale for Neuropsychiatric Symptoms in Pre-Dementia Populations. J Alzheimers Dis 2017;56(03):929-938

MBI has been proposed as a diagnostic category capable of identifying patients at increased risk of developing dementia regardless of the presence of cognitive symptoms. It may help predict the risk of several dementias, including but not exclusive to AD.1717 Ismail Z, Agüera-Ortiz L, Brodaty H, et al; NPS Professional Interest Area of the International Society of to Advance Alzheimer’s Research and Treatment (NPS-PIA of ISTAART) The Mild Behavioral Impairment Checklist (MBI-C): A Rating Scale for Neuropsychiatric Symptoms in Pre-Dementia Populations. J Alzheimers Dis 2017;56(03):929-938 The main rationale for using MBI criteria is the potential to detect neurodegeneration at early stages when therapeutic interventions would be more likely to change disease progression. The pathophysiological processes underlying AD and related dementia start decades before their clinical diagnoses, and although preclinical AD has been recognized as one disease stage in the revised NIA-AA criteria for AD diagnosis,1111 McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011;7(03):263-269,1818 Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011;7(03):280-292 so far, there is no test to predict if someone will develop clinically defined AD. Accordingly, the identification of people with MBI will help define an ‘at high risk’ group for dementia, which is an important step towards the development of effective disease-modifying therapies. Moreover, the treatment of the related behavioral symptoms (as early manifestations of AD) may slow the progression or mitigate the presentation of dementia.1313 Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202 As a word of caution, the diagnostic criteria for MBI represent a significant advance in the definition of behavioral presentations of AD and related dementias, but the concept of MBI still needs to be validated from biological and clinical standpoints.1313 Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202

ASSESSMENT AND MANAGEMENT OF BEHAVIORAL SYMPTOMS IN AD: GENERAL PRINCIPLES

For the assessment of behavioral symptoms in AD, the most used tool is the Neuropsychiatric Inventory (NPI).55 Cummings J. The Neuropsychiatric Inventory: Development and Applications. J Geriatr Psychiatry Neurol 2020;33(02):73-84 Different versions of the NPI can be applied according to the goal or context. In research, the NPI-C, or clinician-version, is frequently used. The clinician scores the behaviors based on the direct assessment of the patient's behavior and taking into account the caregiver and patient's reports. It is more time-consuming but may be less biased than the NPI-Q, which relies only on the partner's report. On the other hand, the NPI-Q is brief and convenient (e.g., can be filled out by the care partner before the appointment), providing an overview of the behaviors presented by the patient and the related impact. Therefore, the NPI-Q may help a busy clinician in their clinical practice.22 Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of Early Alzheimer’s Disease: Clinical Practice in 2021. J Prev Alzheimers Dis 2021;8(03):371-386 Of note, there are other general tools, such as the Behavioral Pathology in Alzheimer's Disease Rating Scale (BEHAVE-AD),1919 Reisberg B, Monteiro I, Torossian C, et al. The BEHAVE-AD assessment system: a perspective, a commentary on new findings, and a historical review. Dement Geriatr Cogn Disord 2014;38;(1-2):89-146 and dedicated instruments for specific behaviors, such as the Cornell Scale for Depression in Dementia,2020 Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell Scale for Depression in Dementia. Biol Psychiatry 1988;23(03):271-284 but they are mainly used in research.

The NPI assesses 12 distinct behaviors that cover the most clinically relevant issues (Table 1). In the past, partly because of the overemphasis on cognitive features of AD, these behaviors were lumped together under umbrella terms like BPSD and NPS and not considered individually.2121 Mathys M. Pharmacologic management of behavioral and psychological symptoms of major neurocognitive disorder. Ment Health Clin 2018;8(06):284-293,2222 Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ 2015;350:h369 Conversely, many of these behaviors actually overlap in clinical practice. Recognizing this overlap, several studies applied statistical methods (e.g., principal component analysis, classic factor analysis, latent class analysis) to reduce the dimensions of behavioral symptoms in AD.2323 Canevelli M, Adali N, Voisin T, et al. Behavioral and psychological subsyndromes in Alzheimer’s disease using the Neuropsychiatric Inventory. Int J Geriatr Psychiatry 2013;28(08):795-803 While there were discrepant results, most studies proposed three to four domains or dimensions that could explain behavioral variability: affective (including apathy, anxiety, depression/dysphoria), psychosis (delusion, hallucination), agitation (aberrant motor behavior, aggression), and disinhibited (elation, disinhibition). These dimensions are clinically relevant as they can guide management and therapeutic interventions.2121 Mathys M. Pharmacologic management of behavioral and psychological symptoms of major neurocognitive disorder. Ment Health Clin 2018;8(06):284-293

Table 1
Behavioral symptoms of Alzheimer's disease

The first step in managing behavioral symptoms in AD is to define the presence and severity of each disruptive behavior. With a defined goal, the second step is to investigate potential causes and/or aggravating factors. These factors can be categorized as patient-, caregiver-, and environment-related2222 Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ 2015;350:h369 (Table 2). Once these factors are addressed (e.g., urinary tract infection treatment, environmental stimuli control), the next stage is to plan the therapeutic interventions. There is a consensus that non-pharmacological approaches are preferred as first-line interventions.2424 Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: an international Delphi consensus. Int Psychogeriatr 2019;31(01):83-90 Discussing these approaches is beyond the scope of this text, but it is important to highlight that many have been evaluated in controlled trials with positive results.2525 Abraha I, Rimland JM, Trotta FM, et al. Systematic review of systematic reviews of non-pharmacological interventions to treat behavioural disturbances in older patients with dementia. The SENATOR-OnTop series. BMJ Open 2017;7(03):e012759 Besides that, these interventions are much safer than pharmacological therapies. A major limitation of non-pharmacological interventions is their access, which may depend on multiple factors, such as the availability of caregivers and/or therapists, and financial resources.

Table 2
Individual-, caregiver- and environment-related factors implicated in the development and/or persistence of behavioral symptoms in Alzheimer's disease

Regarding pharmacological approaches, some general principles must be followed: starting at a low dose, slowly titrating up to the minimum effective dose, and periodically assessing side effects and the need for continued medication prescription.2626 Reus VI, Fochtmann LJ, Eyler AE, et al. The American Psychiatric Association Practice Guideline on the Use of Antipsychotics to Treat Agitation or Psychosis in Patients With Dementia. Am J Psychiatry 2016;173(05):543-546 This is very important because, in contrast to the progressive cognitive decline that defines AD, behavioral symptoms fluctuate during the course of the disease.2727 Borsje P, Wetzels RB, Lucassen PL, Pot AM, Koopmans RT. The course of neuropsychiatric symptoms in community-dwelling patients with dementia: a systematic review. Int Psychogeriatr 2015;27(03):385-405,2828 van der Linde RM, Dening T, Stephan BC, Prina AM, Evans E, Brayne C. Longitudinal course of behavioural and psychological symptoms of dementia: systematic review. Br J Psychiatry 2016; 209(05):366-377 The decision on the specific medication must consider the targeted behavioral symptom or domain, the patient's medical comorbidities, and polypharmacy.2929 Tampi RR, Jeste DV. Dementia Is More Than Memory Loss: Neuropsychiatric Symptoms of Dementia and Their Nonpharmacological and Pharmacological Management. Am J Psychiatry 2022;179(08):528-543,3030 Caramelli P, Marinho V, Laks J, et al. Treatment of dementia: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022;16(03, Suppl 1):88-100 For instance, for affective symptoms, antidepressants, mainly selective serotonin reuptake inhibitors (SSRIs), are the primary choice. Optimization of anticholinesterase treatment seems to contribute to these symptoms as well. Emerging evidence has indicated that this class of medication is helpful not only in managing cognitive and other behavioral symptoms but also positively affects dementia-related outcomes (i.e., risk of death or severe dementia).3131 Tan ECK, Johnell K, Bell JS, et al. Do Acetylcholinesterase Inhibitors Prevent or Delay Psychotropic Prescribing in People With Dementia? Analyses of the Swedish Dementia Registry. Am J Geriatr Psychiatry 2020;28(01):108-1173333 Marucci G, Buccioni M, Ben DD, Lambertucci C, Volpini R, Amenta F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology 2021;190:108352 For agitation, antidepressants can also be tried, while atypical antipsychotics should be reserved for severe cases and used after weighing the associated cardiovascular and death risks (see below).

The contemporary trend is to define a Diagnostic and Statistical Manual (DSM)-like set of criteria for specific behaviors, such as apathy, agitation, and psychosis, as it will be discussed in the following sections.

APATHY, DEPRESSION, AND ANXIETY

Apathy is a behavioral syndrome defined by reduced or loss of motivation for goal-directed behaviors and cognitive activities alongside blunted affect.3434 Teixeira AL, Gonzales MM, de Souza LC, Weisenbach SL. Revisiting Apathy in Alzheimer’s Disease: From Conceptualization to Therapeutic Approaches. Behav Neurol 2021;2021:6319826 Apathy is the most common behavioral disorder in AD, with a prevalence of up to 90% in severe dementia. It has been associated with a series of negative clinical outcomes, including cognitive and functional decline, caregiver burden, institutionalization, and mortality, explaining a significant portion of the impact of behavioral or neuropsychiatric symptoms in AD.3434 Teixeira AL, Gonzales MM, de Souza LC, Weisenbach SL. Revisiting Apathy in Alzheimer’s Disease: From Conceptualization to Therapeutic Approaches. Behav Neurol 2021;2021:6319826 Among all behavioral symptoms, apathy is the one closer correlated with cognitive decline in AD, predicting progression from normal cognition to MCI and from MCI to dementia.3535 Salem H, Suchting R, Gonzales MM, Seshadri S, Teixeira AL. Apathy as a Predictor of Conversion from Mild Cognitive Impairment to Alzheimer’s Disease: A Texas Alzheimer’s Research and Care Consortium (TARCC) Cohort-Based Analysis. J Alzheimers Dis 2023;92(01):129-139

The International Society for CNS Clinical Trials and Methodology and the European Psychiatric Association proposed diagnostic criteria for apathy in neurocognitive disorders within a framework similar to the DSM. Accordingly, an individual with AD is diagnosed as apathetic when they meet four criteria (A-D).3636 Miller DS, Robert P, Ereshefsky L, et al. Diagnostic criteria for apathy in neurocognitive disorders. Alzheimers Dement 2021;17(12):1892-1904 Criterion A requires the diagnosis of cognitive impairment or dementia. Criterion B requires the presence of at least one persistent or recurrent symptom in at least two of three domains (i.e., diminished initiative, diminished interest, diminished emotional expression/responsiveness) for a minimum of four weeks. For example, a patient may be less likely to spontaneously initiate usual activities, such as hobbies, less interested in participating in everyday activities, and emotionally blunted. Criterion C is exclusionary and postulates that these symptoms are not solely explained by physical or motor disabilities, impaired arousal, the effect of a substance, or a psychiatric disorder. Criterion D states that these symptoms cause significant impairment in personal, social, occupational, and other areas of functioning.

Apathy has been conceptualized as a transdiagnostic syndrome within the disorders of motivation and reward.3434 Teixeira AL, Gonzales MM, de Souza LC, Weisenbach SL. Revisiting Apathy in Alzheimer’s Disease: From Conceptualization to Therapeutic Approaches. Behav Neurol 2021;2021:6319826,3737 Husain M, Roiser JP. Neuroscience of apathy and anhedonia: a transdiagnostic approach. Nat Rev Neurosci 2018;19(08):470-484 Dopaminergic circuits play a major role in this context, thus pharmacological approaches activating dopamine signaling have been used for apathy. Methylphenidate was shown to be an effective treatment for apathy in AD in open and controlled studies but with concerns related to potential weight loss, increased anxiety, and cardiovascular effects.3838 Sassi KLM, Rocha NP, Colpo GD, John V, Teixeira AL. Amphetamine Use in the Elderly: A Systematic Review of the Literature. Curr Neuropharmacol 2020;18(02):126-135,3939 Mintzer J, Lanctôt KL, Scherer RW, et al; ADMET 2 Research Group. Effect of Methylphenidate on Apathy in Patients With Alzheimer Disease: The ADMET 2 Randomized Clinical Trial. JAMA Neurol 2021;78(11):1324-1332

There is considerable overlap (30-50%) between apathy and depression in AD.3434 Teixeira AL, Gonzales MM, de Souza LC, Weisenbach SL. Revisiting Apathy in Alzheimer’s Disease: From Conceptualization to Therapeutic Approaches. Behav Neurol 2021;2021:6319826,4040 Burke AD, Goldfarb D, Bollam P, Khokher S. Diagnosing and Treating Depression in Patients with Alzheimer’s Disease. Neurol Ther 2019;8(02):325-350 Depression is also a very common behavioral problem in AD, with an estimated prevalence of major depression in AD around 15%, with higher numbers when milder forms of depressive disorders are considered.4141 Asmer MS, Kirkham J, Newton H, et al. Meta-Analysis of the Prevalence of Major Depressive Disorder Among Older Adults With Dementia. J Clin Psychiatry 2018;79(05):17r11772 As the lack of motivation is one of the DSM criteria for the diagnosis of major depression, the differential diagnosis between apathy and depression can be challenging.4040 Burke AD, Goldfarb D, Bollam P, Khokher S. Diagnosing and Treating Depression in Patients with Alzheimer’s Disease. Neurol Ther 2019;8(02):325-350 In clinical practice, depression can be differentiated from apathy as the former involves subjective feelings of sadness, negative thoughts (e.g., guilt, hopelessness, low self-esteem), and sleep disorders (e.g., early awakening).

There are National Institute of Mental Health (NIMH)-sponsored diagnostic criteria for depression in AD based on the DSM criteria for major depression.4242 Olin JT, Schneider LS, Katz IR, et al. Provisional diagnostic criteria for depression of Alzheimer disease. Am J Geriatr Psychiatry 2002;10(02):125-128 The criteria for depression in AD require the presence of three – at least one must be depressed mood or decreased positive affect/pleasure in response to usual activities – out of ten symptoms (depressed mood, decreased positive affect or pleasure, social isolation, disruption in appetite, disruption in sleep, psychomotor changes, irritability, fatigue, recurrent thoughts of death or suicide, and feelings of worthlessness, hopelessness or guilt) during a two-week period, with a definite change from the previous level of functioning. In addition to the NPI, the Cornell Scale for Depression in Dementia has been used to assess depression in AD.2020 Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell Scale for Depression in Dementia. Biol Psychiatry 1988;23(03):271-284 As it is a clinician-rated scale considering both patient and caregiver perspectives, the Cornell Scale for Depression in Dementia can better control biased information related to anosognosia, normalization of symptoms, and/or caregiver stress.4343 Agüera-Ortiz L, García-Ramos R, Grandas Pérez FJ, et al. Depression in Alzheimer’s Disease: A Delphi Consensus on Etiology, Risk Factors, and Clinical Management. Front Psychiatry 2021;12:638651 Pharmacological therapy for depression in AD usually relies on SSRIs (e.g., sertraline, citalopram),4040 Burke AD, Goldfarb D, Bollam P, Khokher S. Diagnosing and Treating Depression in Patients with Alzheimer’s Disease. Neurol Ther 2019;8(02):325-350 with a growing tendency to use dual antidepressants, such as duloxetine, venlafaxine/desvenlafaxine.4343 Agüera-Ortiz L, García-Ramos R, Grandas Pérez FJ, et al. Depression in Alzheimer’s Disease: A Delphi Consensus on Etiology, Risk Factors, and Clinical Management. Front Psychiatry 2021;12:638651 It is worth mentioning that antidepressants may worsen apathy.3434 Teixeira AL, Gonzales MM, de Souza LC, Weisenbach SL. Revisiting Apathy in Alzheimer’s Disease: From Conceptualization to Therapeutic Approaches. Behav Neurol 2021;2021:6319826

While apathy and depression have been relatively well investigated in AD, much less is known about anxiety.4444 Mendez MF. The Relationship Between Anxiety and Alzheimer’s Disease. J Alzheimers Dis Rep 2021;5(01):171-177 Anxiety is the third most common behavioral disorder in AD, with an estimated prevalence of around 40%.44 Zhao QF, Tan L, Wang HF, et al. The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: Systematic review and meta-analysis. J Affect Disord 2016;190:264-271 In the early stages of AD, anxiety resembles its presentation in adults, i.e., characterized by worries and somatic symptoms, while in later phases of the disease, it manifests as emotional distress, irritability, and excessive motor activity. Moreover, anxiety tends to aggregate with depression within an affective behavioral domain but is less associated with cognitive decline in AD than depression and apathy. The management of anxiety in AD emphasizes non-pharmacological strategies (e.g., physical activity, music, occupational and cognitive therapies), especially given the significant risks of using benzodiazepines in older adults, such as cognitive decline, falls, and sedation. SSRIs are the drugs of choice for anxiety in AD, and trazodone can be considered when associated with insomnia.4444 Mendez MF. The Relationship Between Anxiety and Alzheimer’s Disease. J Alzheimers Dis Rep 2021;5(01):171-177

PSYCHOSIS

Psychosis is a descriptive term encompassing delusions and hallucinations.4545 Cummings J, Pinto LC, Cruz M, et al. Criteria for Psychosis in Major and Mild Neurocognitive Disorders: International Psychogeriatric Association (IPA) Consensus Clinical and Research Definition. Am J Geriatr Psychiatry 2020;28(12):1256-1269,4646 Sano M, Cummings J, Jeste DV, Finkel S, Reichman W. International Psychogeriatric Association (IPA) consensus for defining psychosis in major and mild neurocognitive disorders. Int Psychogeriatr 2022;34(03):203-207 Psychosis is a clinically relevant neuropsychiatric syndrome reported in 41% of patients with AD, with delusions in 36% and hallucinations in 18%.4747 Ropacki SA, Jeste DV. Epidemiology of and risk factors for psychosis of Alzheimer’s disease: a review of 55 studies published from 1990 to 2003. Am J Psychiatry 2005;162(11):2022-2030

From a psychopathological perspective, there are different types of delusions (e.g., persecutory, religiosity, misidentification) and hallucinations (e.g., visual, auditory, somatic) in AD. Furthermore, they may differ during the course of the disease. Paranoid or persecutory delusions, including delusions of theft or infidelity, tend to occur earlier than misidentification delusions.4848 Pearce D, Gould RL, Roughley M, et al. Paranoid and misidentification subtypes of psychosis in dementia. Neurosci Biobehav Rev 2022;134:104529 The latter delusions are commonly associated with more severe cognitive impairments, mainly perceptual deficits (e.g., prosopagnosia), and include Capgras (or hypofamiliarity), in which the patient firmly believes that a relative or a friend is an impostor, Fregoli (or hyperfamiliarity), convinced that a close person constantly changes their appearance or incarnate in others, phantom boarder syndromes, the belief that uninvited people are living in the house, and reduplicative paramnesias, the belief that a place a familiar place exists in two or more physical locations simultaneously.4848 Pearce D, Gould RL, Roughley M, et al. Paranoid and misidentification subtypes of psychosis in dementia. Neurosci Biobehav Rev 2022;134:104529 Hallucinations often present with delusions rather than alone, frequently overlapping with them, such as misidentification syndromes. Somatic hallucinations are also difficult to disentangle from somatic delusions, as in Ekbom syndrome or delusional parasitosis.4949 Nicolato R, Corrêa H, Romano-Silva MA, Teixeira AL Jr. Delusional parasitosis or Ekbom syndrome: a case series. Gen Hosp Psychiatry 2006;28(01):85-87 Visual hallucinations are the most frequent, and hallucinatory phenomena can range from simple (e.g., unformed images, noises) to complex characteristics (e.g., people, conversations).

Psychosis tends to persist for months,4747 Ropacki SA, Jeste DV. Epidemiology of and risk factors for psychosis of Alzheimer’s disease: a review of 55 studies published from 1990 to 2003. Am J Psychiatry 2005;162(11):2022-2030 and this protracted course helps to discriminate it from delirium, which usually has an acute or subacute presentation. It is also important to distinguish delusions from confabulations, i.e., the fabrication of false or distorted memories. Psychosis can be distressing to the patient, leading to agitation and/or aggression, especially when confronted by a caregiver or a family member. Acknowledging these features is important to establish a non-pharmacological approach that includes a non-confrontational or argumentative attitude while attempting to ensure the patient's safety.

Antipsychotics have a modest effect on the treatment of psychotic symptoms in AD. Still, this effect comes at the cost of potentially harmful side effects, including cognitive, motor, and cardiovascular events and increased risk of death.5050 Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353(22):2335-23415353 Vigen CL, Mack WJ, Keefe RS, et al. Cognitive effects of atypical antipsychotic medications in patients with Alzheimer’s disease: outcomes from CATIE-AD. Am J Psychiatry 2011;168(08):831-839 As typical antipsychotics (e.g., haloperidol, chlorpromazine) are associated with higher rates of side effects compared to atypical ones, most of the literature has focused on the latter. Meta-analytical evidence of atypical antipsychotics has shown small but statistically significant benefits with aripiprazole, olanzapine, and risperidone compared to quetiapine.5454 Maher AR, Maglione M, Bagley S, et al. Efficacy and comparative effectiveness of atypical antipsychotic medications for off-label uses in adults: a systematic review and meta-analysis. JAMA 2011;306(12):1359-1369 A network meta-analysis found that aripiprazole was the most effective and safe atypical antipsychotic for behavioral symptoms – as assessed by total scores in general psychopathology tools, not only psychosis domains – olanzapine provided the least benefit overall, while risperidone and quetiapine had intermediate profiles of effectiveness and safety.5555 Yunusa I, Alsumali A, Garba AE, Regestein QR, Eguale T. Assessment of Reported Comparative Effectiveness and Safety of Atypical Antipsychotics in the Treatment of Behavioral and Psychological Symptoms of Dementia: A Network Meta-analysis. JAMA Netw Open 2019;2(03):e190828 However, a more recent meta-analysis of atypical antipsychotics (quetiapine, risperidone, olanzapine, aripiprazole, and brexpiprazole) for the treatment of AD-related psychosis failed to show any statistically significant improvement in symptoms, but all were associated with greater odds of mortality.5656 Yunusa I, Rashid N, Demos GN, Mahadik BS, Abler VC, Rajagopalan K. Comparative Outcomes of Commonly Used Off-Label Atypical Antipsychotics in the Treatment of Dementia-Related Psychosis: A Network Meta-analysis. Adv Ther 2022;39(05): 1993-2008 Despite their risk and effectiveness controversies, approximately one-third of patients with dementia use antipsychotics, with marked variation according to care settings and severity of disease.5757 Kirkham J, Sherman C, Velkers C, et al. Antipsychotic Use in Dementia. Can J Psychiatry 2017;62(03):170-181 Community-based studies have shown a much lower frequency of antipsychotic use when compared with long-term care settings (12.3% vs. 37.5%), while the prevalence of antipsychotic use increases from 6.7% in mild to 12.2% in moderate and up to 45.1% in severe dementia.5757 Kirkham J, Sherman C, Velkers C, et al. Antipsychotic Use in Dementia. Can J Psychiatry 2017;62(03):170-181

In this scenario, alternative pharmacological strategies for psychosis have been tested. Pimavanserin is a highly selective serotonin 5-HT2A receptor inverse agonist and antagonist with minimal dopaminergic, histaminergic, and muscarinic effects. This unique profile drug has shown to be effective for psychosis in Parkinson's disease, being FDA-approved for this indication.5858 Cummings J, Isaacson S, Mills R, et al. Pimavanserin for patients with Parkinson’s disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet 2014;383(9916):533-540 Tariot et al.5959 Tariot PN, Cummings JL, Soto-Martin ME, et al. Trial of Pimavanserin in Dementia-Related Psychosis. N Engl J Med 2021;385(04):309-319 conducted a phase 3, double-blind, randomized, placebo-controlled discontinuation trial with patients with psychosis related to AD and other non-AD dementias. Patients received open-label pimavanserin for 12 weeks. Those with a reduction from baseline of at least 30% in psychotic symptoms were randomly assigned at week 12 in a 1:1 ratio to continue receiving pimavanserin or to receive a placebo for up to 26 weeks. A relapse occurred in 12 of 95 patients (13%) in the pimavanserin group and in 28 of 99 (28%) in the placebo group (hazard ratio, 0.35; P = 0.005), favoring the former. Side effects occurred in 41% of the pimavanserin group and included headache, constipation, urinary tract infection, and asymptomatic QT interval prolongation. Despite promising efficacy results and an acceptable side-effect profile, the study was underpowered to show significant effects in specific types of dementia. In our experience, pimavanserin can be used in AD patients with less severe psychosis, but it should not be used in association with atypical antipsychotics because of safety concerns.6060 Moreno GM, Gandhi R, Lessig SL, Wright B, Litvan I, Nahab FB. Mortality in patients with Parkinson disease psychosis receiving pimavanserin and quetiapine. Neurology 2018;91(17):797-799

Finally, it is worth highlighting marked advances in the understanding of the neurobiology of psychosis in AD, as reviewed elsewhere.6161 Ismail Z, Creese B, Aarsland D, et al. Psychosis in Alzheimer disease - mechanisms, genetics and therapeutic opportunities. Nat Rev Neurol 2022;18(03):131-144 Besides the overlap between the genetic architecture of AD-related psychosis and primary psychiatric disorders presenting with psychosis (schizophrenia, bipolar disorder), reinforcing the transdiagnostic validity of psychosis, studies have shown the contribution of Lewy body and vascular pathology.

AGITATION

Agitation is a very troublesome behavioral problem in AD with major impacts, including costs and caregiver burden.6262 Sano M, Cummings J, Auer S, et al. Agitation in cognitive disorders: Progress in the International Psychogeriatric Association consensus clinical and research definition. Int Psychogeriatr 2023; •••:1-13,6363 Cummings J, Sano M, Auer S, et al. Reduction and prevention of agitation in persons with neurocognitive disorders: an international psychogeriatric association consensus algorithm. Int Psychogeriatr 2023;•••:1-12 In a community-dwelling study involving older adults with a range of cognitive impairments, the prevalence of agitation ranged from 8.3% to 48.9%.6464 Sano M, Zhu CW, Neugroschl J, Grossman HT, Schimming C, Aloysi A. Agitation in Cognitive Disorders: Use of the National Alzheimer’s Coordinating Center Uniform Data Set (NACC-UDS) to Evaluate International Psychogeriatric Association Definition. Am J Geriatr Psychiatry 2022;30(11):1198-1208 Agitation increases with dementia severity, and even higher rates (> 50%) are observed in nursing homes.6565 Selbaek G, Engedal K, Benth JŠ, Bergh S. The course of neuropsychiatric symptoms in nursing-home patients with dementia over a 53-month follow-up period. Int Psychogeriatr 2014;26(01):81-91

The International Psychogeriatric Association consensus panel considered not only excessive motor activity (e.g., restlessness, pacing, repetitive mannerisms) but also verbal (e.g., yelling, using profanities) and physical (e.g., hitting, scratching, biting, throwing objects) aggression under the definition of agitation.6262 Sano M, Cummings J, Auer S, et al. Agitation in cognitive disorders: Progress in the International Psychogeriatric Association consensus clinical and research definition. Int Psychogeriatr 2023; •••:1-13 Although agitation is not a synonym for aggression, the panel acknowledged that most studies do not discriminate against these behaviors and that this position also reflects how clinicians and families address the problem. According to the same criteria, these behaviors should be intermittently or persistently present for at least two weeks, which excludes acute agitation. Acute agitation must prompt careful investigation of environmental (e.g., movement to an unfamiliar palace) and medical causes (e.g., physical distress, pain, medication-related, urinary tract infection), ruling out agitated delirium.

Two different types of aggression – reactive and proactive – have been recognized.6262 Sano M, Cummings J, Auer S, et al. Agitation in cognitive disorders: Progress in the International Psychogeriatric Association consensus clinical and research definition. Int Psychogeriatr 2023; •••:1-13 The former seems to be more frequent in the context of dementia, while the second is mainly associated with psychotic phenomena. In line with this difference, it is worth noticing that agitation and psychosis usually do not load on the same behavioral domains (see above: Assessment and management of behavioral symptoms of AD). The relationship between agitation and other behavioral symptoms, such as affective and disinhibition/impulsive, tends to follow the same pattern, as they tend to load in discrete domains.2323 Canevelli M, Adali N, Voisin T, et al. Behavioral and psychological subsyndromes in Alzheimer’s disease using the Neuropsychiatric Inventory. Int J Geriatr Psychiatry 2013;28(08):795-803 Another important clinical aspect is the definition of whether agitation has a circadian rhythm, such as in ‘sundowning’ and nighttime disruptive behaviors.

Recently, the International Psychogeriatrics (IPA) Agitation Work Group proposed an algorithm for managing agitation in neurocognitive disorders.6363 Cummings J, Sano M, Auer S, et al. Reduction and prevention of agitation in persons with neurocognitive disorders: an international psychogeriatric association consensus algorithm. Int Psychogeriatr 2023;•••:1-12 Using the ‘Investigate, Plan, and Act’ (also IPA) approach, in which the investigation of potential causative or contributor factors is followed by planning and acting, with reiterations of the process until agitation is controlled, the Working Group emphasized the integration of psychosocial and pharmacologic interventions. Psychosocial interventions that have multiple components target the caregiver (e.g., communication skills, education, self-care) and the patient (e.g., person-centered, aerobic activities, music, massage) and are delivered with regular follow-up have more therapeutic benefit.6666 Livingston G, Kelly L, Lewis-Holmes E, et al. Non-pharmacological interventions for agitation in dementia: systematic review of randomised controlled trials. Br J Psychiatry 2014;205(06):436-442,6767 Watt JA, Goodarzi Z, Veroniki AA, et al. Comparative Efficacy of Interventions for Aggressive and Agitated Behaviors in Dementia: A Systematic Review and Network Meta-analysis. Ann Intern Med 2019;171(09):633-642 Pharmacological treatment should be considered only after psychosocial interventions failed to reduce agitation. Before adding any new medication, it is fundamental to review the current regimen in search of drugs that can cause or aggravate agitation. For instance, benzodiazepines, opiates and anticholinergic drugs can cause delirium. Dopamine-blocking agents can cause akathisia, a condition characterized by subjective and objective restlessness and typically seen in patients taking antipsychotics.6868 Salem H, Nagpal C, Pigott T, Teixeira AL. Revisiting Antipsychotic-induced Akathisia: Current Issues and Prospective Challenges. Curr Neuropharmacol 2017;15(05):789-798

For nighttime disruptive behaviors, trazodone and orexin receptor antagonists (e.g., suvorexant, lemborexant) have been used [63]. Of note, a recent double-blind, placebo-controlled study (SYMBAD) found no effect of mirtazapine – an atypical antidepressant with sedative properties at lower doses but activating in higher – on AD-related agitation and potentially increased mortality associated with its use.6969 Banerjee S, High J, Stirling S, et al. Study of mirtazapine for agitated behaviours in dementia (SYMBAD): a randomised, double-blind, placebo-controlled trial. Lancet 2021;398(10310):1487-1497 Actually, antidepressants can be used as the first-line pharmacological strategy given the emotional distress usually underlying agitation. Citalopram is the most well-studied antidepressant and has been shown to reduce agitation in AD, especially those with mild to moderate agitation and less cognitive impairment.7070 Porsteinsson AP, Drye LT, Pollock BG, et al; CitAD Research Group. Effect of citalopram on agitation in Alzheimer disease: the CitAD randomized clinical trial. JAMA 2014;311(07):682-691,7171 Schneider LS, Frangakis C, Drye LT, et al; CitAD Research Group. Heterogeneity of Treatment Response to Citalopram for Patients With Alzheimer’s Disease With Aggression or Agitation: The CitAD Randomized Clinical Trial. Am J Psychiatry 2016;173(05):465-472 As discussed in the previous section, atypical antipsychotics should be reserved for more severe cases. Risperidone is approved for agitation in dementia in some countries (e.g., Australia, Canada, United Kingdom), and brexpiprazole was recently approved in the United States. Actually, brexpiprazole was the first Food Drug Administration (FDA)-approved drug for an AD-related behavioral symptom, i.e., agitation.7272 Administration, F.a.D. FDA Approves First Drug to Treat Agitation Symptoms Associated with Dementia due to Alzheimer's Disease. 2023 05/11/2023 [cited 2023. The FDA granted a ‘Fast Track’ designation – that addresses a serious condition and fills an unmet medical need – to the application consisting of two 12-week, randomized, double-blind, placebo-controlled, fixed-dose studies. In the first study, patients received 1 or 2 mg, while patients received 2 or 3 mg in the second study, with the primary efficacy endpoint being the change from baseline in the Cohen-Mansfield Agitation Inventory total score at week 12. Patients who received 2 mg or 3 mg of brexpiprazole had clinically meaningful improvements compared to the placebo group. This approval reflects, at least in part, the significant unmet needs in the management of behavioral symptoms in AD and the related societal pressure. In the case of partial or non-response with antipsychotics, the mood-stabilizing drugs carbamazepine, gabapentin, and valproic acid should be considered.7373 Davies SJ, Burhan AM, Kim D, et al. Sequential drug treatment algorithm for agitation and aggression in Alzheimer's and mixed dementia. J Psychopharmacol 2018;32(05):509-523,7474 Supasitthumrong T, Bolea-Alamanac BM, Asmer S, Woo VL, Abdool PS, Davies SJC. Gabapentin and pregabalin to treat aggres- sivity in dementia: a systematic review and illustrative case report. Br J Clin Pharmacol 2019;85(04):690-703

Emerging therapeutics for agitation in AD include pimavanserin (discussed above), dextromethorphan/quinidine, and cannabinoids. Dextromethorphan/quinidine is an FDA-approved drug combination for the treatment of pseudobulbar affect (or pathological laughing/crying, involuntary emotional expression disorder) associated with chronic neurological conditions, such as multiple sclerosis and amyotrophic lateral sclerosis. In a randomized phase 2, double-blind, placebo-controlled trial, dextromethorphan/quinidine demonstrated clinically relevant efficacy for agitation and was well tolerated.7575 Cummings JL, Lyketsos CG, Peskind ER, et al. Effect of Dextrome-thorphan-Quinidine on Agitation in Patients With Alzheimer Disease Dementia: A Randomized Clinical Trial. JAMA 2015;314(12):1242-1254 In a randomized double-blind crossover trial comparing nabilone – a synthetic tetrahydrocannabinol analog – with a placebo, nabilone was effective for agitation but caused sedation and cognitive impairment.7676 Herrmann N, Ruthirakuhan M, Gallagher D, et al. Randomized Placebo-Controlled Trial of Nabilone for Agitation in Alzheimer’s Disease. Am J Geriatr Psychiatry 2019;27(11):1161-1173 A meta-analysis of the literature including natural and synthetic cannabinoids for agitation in AD concluded there may be a signal for a potential benefit of synthetic cannabinoids, but there are concerns with side effects, mainly sedation.7777 Ruthirakuhan M, Lanctôt KL, Vieira D, Herrmann N. Natural and Synthetic Cannabinoids for Agitation and Aggression in Alzheimer’s Disease: A Meta-Analysis. J Clin Psychiatry 2019;80(02):18r12617

In sum, no pharmacological interventions for agitation in AD have therapeutic benefits that clearly outweigh their potential side effects and related safety concerns.7878 Herrmann N, Wang HJ, Song BX, Bawa KK, Lanctôt KL. Risks and benefits of current and novel drugs to treat agitation in Alzheimer’s disease. Expert Opin Drug Saf 2022;21(10):1289-1301 Non-pharmacological or psychosocial interventions should be regarded as the mainstay of the treatment of agitation in AD.

SLEEP DISORDERS

There is a bidirectional relationship between AD and sleep disorders. In older individuals, sleep disorders are risk factors for cognitive impairment and dementia.7979 Shi L, Chen SJ, Ma MY, et al. Sleep disturbances increase the risk of dementia: A systematic review and meta-analysis. Sleep Med Rev 2018;40:4-16,8080 Bubu OM, Brannick M, Mortimer J, et al. Sleep, Cognitive impairment, and Alzheimer’s disease: A Systematic Review and Meta-Analysis. Sleep 2017;40(01): Sleep disorders have also been identified as a predictor of dementia incidence in prodromal phases of AD, such as MCI,8181 Casagrande M, Forte G, Favieri F, Corbo I. Sleep Quality and Aging: A Systematic Review on Healthy Older People, Mild Cognitive Impairment and Alzheimer’s Disease. Int J Environ Res Public Health 2022;19(14):8457 with an estimated population attributable risk of AD because of sleep disorders around 15%.8080 Bubu OM, Brannick M, Mortimer J, et al. Sleep, Cognitive impairment, and Alzheimer’s disease: A Systematic Review and Meta-Analysis. Sleep 2017;40(01): Conversely, the prevalence of sleep disorders among individuals with AD ranges from 14% to 69%, with a pooled prevalence estimate of 39% (95% CI 30–47%).44 Zhao QF, Tan L, Wang HF, et al. The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: Systematic review and meta-analysis. J Affect Disord 2016;190:264-271 The most commonly reported sleep disorders in AD are insomnia, sleep-disordered breathing (SDB), restless legs syndrome, and rapid eye movement (REM) sleep behavior disorder.8181 Casagrande M, Forte G, Favieri F, Corbo I. Sleep Quality and Aging: A Systematic Review on Healthy Older People, Mild Cognitive Impairment and Alzheimer’s Disease. Int J Environ Res Public Health 2022;19(14):8457

While sleep disorders may contribute to AD pathophysiology, AD-related pathology contributes to sleep problems.8181 Casagrande M, Forte G, Favieri F, Corbo I. Sleep Quality and Aging: A Systematic Review on Healthy Older People, Mild Cognitive Impairment and Alzheimer’s Disease. Int J Environ Res Public Health 2022;19(14):8457 Sleep disorders can exacerbate cognitive symptoms by impairing sleep-dependent memory consolidation processes.8080 Bubu OM, Brannick M, Mortimer J, et al. Sleep, Cognitive impairment, and Alzheimer’s disease: A Systematic Review and Meta-Analysis. Sleep 2017;40(01): Degeneration of neural pathways that regulate sleep-wake cycles and sleep architecture, such as the suprachiasmatic nucleus, can lead to sleep-wake cycle disorders, resulting in fragmented sleep and sleep-wake cycle reversal, with daytime sleepiness and nocturnal agitation. Medications used to treat AD-related symptoms can further contribute to sleep dysfunction. For example, cholinesterase inhibitors can cause vivid dreams and nightmares, while antipsychotic medications can cause sedation.8282 Javed B, Javed A, Kow CS, Hasan SS. Pharmacological and non-pharmacological treatment options for sleep disturbances in Alzheimer’s disease. Expert Rev Neurother 2023;23(06):501-514

In this context, sleep disorders are highly prevalent among individuals with AD, and their presence worsens disease presentation and may accelerate disease progression. Yet, their management in AD is very limited. Non-pharmacological interventions (e.g., exercise, continuous positive airway pressure (CPAP), bright light therapy) are the first-line approach for sleep problems in AD, as they conciliate a favorable safety profile with reported efficacy.8282 Javed B, Javed A, Kow CS, Hasan SS. Pharmacological and non-pharmacological treatment options for sleep disturbances in Alzheimer’s disease. Expert Rev Neurother 2023;23(06):501-514 When these interventions do not work, pharmacological strategies can be considered. There is a dearth of evidence of the efficacy of many drugs widely prescribed for sleep disorders in dementia, including benzodiazepine and non-benzodiazepine hypnotics (e.g., zolpidem, eszopiclone). Moreover, the use of these drugs is associated with potentially meaningful side effects, such as risk of falls and cognitive decline. Melatonin is widely used for treating sleep problems in AD, especially due to its positive safety profile and potential to regulate disrupted circadian rhythms. However, the evidence of melatonin efficacy in improving sleep disorders in AD is limited. Although a randomized, placebo-controlled trial showed a significant improvement in subjective sleep measures with melatonin compared to placebo,8383 Wade AG, Farmer M, Harari G, et al. Add-on prolonged-release melatonin for cognitive function and sleep in mild to moderate Alzheimer’s disease: a 6-month, randomized, placebo-controlled, multicenter trial. Clin Interv Aging 2014;9:947-961 others found no clinical benefit of melatonin when sleep outcomes were measured objectively (e.g., actigraphy).8484 Serfaty M, Kennell-Webb S, Warner J, Blizard R, Raven P. Double blind randomised placebo controlled trial of low dose melatonin for sleep disorders in dementia. Int J Geriatr Psychiatry 2002;17 (12):1120-1127,8585 Gehrman PR, Connor DJ, Martin JL, Shochat T, Corey-Bloom J, Ancoli-Israel S. Melatonin fails to improve sleep or agitation in double-blind randomized placebo-controlled trial of institutionalized patients with Alzheimer disease. Am J Geriatr Psychiatry 2009;17(02):166-169 The most recent Cochrane Review on the topic concluded that there is no evidence of the efficacy of melatonin (up to 10 mg) or melatonin receptor agonists (e.g., agomelatine, ramelteon) for sleep problems in AD (including total nocturnal sleep time, mean duration of sleep bouts, sleep efficiency, number of nocturnal awakenings, and sleep latency).8686 McCleery J, Sharpley AL. Pharmacotherapies for sleep disturbances in dementia. Cochrane Database Syst Rev 2020;11(11):CD009178 There is evidence of potential beneficial effects on sleep outcomes in AD, with a relatively safe profile, for trazodone and orexin antagonists (e.g., suvorexant).8686 McCleery J, Sharpley AL. Pharmacotherapies for sleep disturbances in dementia. Cochrane Database Syst Rev 2020;11(11):CD009178 There is a clear inconsistency between the current evidence versus the clinical practice to treat sleep disorders in AD. A systematic assessment of the benefits and adverse effects of drugs used to treat sleep problems in AD is urgently needed.

In conclusion, in addition to its core clinical features of cognitive and functional decline, AD is characterized by a series of neuropsychiatric or behavioral symptoms. These symptoms most commonly include apathy, depression, anxiety, agitation, and psychosis, which significantly contribute to the clinical heterogeneity of AD and, thus, to its challenging management. Indeed, they are often debilitating, impacting several outcomes, such as caregiver burden and service utilization. Several non-pharmacological approaches for behavioral symptoms in AD have been proposed and demonstrated to be effective, but there are barriers to their implementation and they may not be enough to control them. Pharmacological treatments have shown modest efficacy, but potentially serious side effects, especially related to antipsychotics. Furthermore, the dynamic nature of these symptoms, which tend to fluctuate in frequency and severity, requires constant reassessment and flexibility of the therapeutic plan. There is a great need to develop safer and more efficacious strategies for the management of behavioral symptoms of AD.

Acknowledgements

The Neuropsychiatry Program (ALT) is supported by the Texas Alzheimer's Research and Care Consortium (TARCC 2020-23-93-II) and NIH/NIA R21 AG072380-01A1. JG is supported by NIH/NIA K23 AG058805 and NIH/NIA R01 AG078191.

References

  • 1
    Scheltens P, De Strooper B, Kivipelto M, et al. Alzheimer’s disease. Lancet 2021;397(10284):1577-1590
  • 2
    Porsteinsson AP, Isaacson RS, Knox S, Sabbagh MN, Rubino I. Diagnosis of Early Alzheimer’s Disease: Clinical Practice in 2021. J Prev Alzheimers Dis 2021;8(03):371-386
  • 3
    Steinberg M, Shao H, Zandi P, et al; Cache County Investigators. Point and 5-year period prevalence of neuropsychiatric symptoms in dementia: the Cache County Study. Int J Geriatr Psychiatry 2008;23(02):170-177
  • 4
    Zhao QF, Tan L, Wang HF, et al. The prevalence of neuropsychiatric symptoms in Alzheimer’s disease: Systematic review and meta-analysis. J Affect Disord 2016;190:264-271
  • 5
    Cummings J. The Neuropsychiatric Inventory: Development and Applications. J Geriatr Psychiatry Neurol 2020;33(02):73-84
  • 6
    Cummings J. The Role of Neuropsychiatric Symptoms in Research Diagnostic Criteria for Neurodegenerative Diseases. Am J Geriatr Psychiatry 2021;29(04):375-383
  • 7
    Cerejeira J, Lagarto L, Mukaetova-Ladinska EB. Behavioral and psychological symptoms of dementia. Front Neurol 2012;3:73
  • 8
    Benjamin S, MacGillivray L, Schildkrout B, Cohen-Oram A, Lauterbach MD, Levin LL. Six Landmark Case Reports Essential for Neuropsychiatric Literacy. J Neuropsychiatry Clin Neurosci 2018;30(04):279-290
  • 9
    Feast A, Moniz-Cook E, Stoner C, Charlesworth G, Orrell M. A systematic review of the relationship between behavioral and psychological symptoms (BPSD) and caregiver well-being. Int Psychogeriatr 2016;28(11):1761-1774
  • 10
    Sabates J, Chiu WH, Loi S, et al. The Associations Between Neuropsychiatric Symptoms and Cognition in People with Dementia: A Systematic Review and Meta-Analysis. Neuropsychol Rev 2023
  • 11
    McKhann GM, Knopman DS, Chertkow H, et al. The diagnosis of dementia due to Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011;7(03):263-269
  • 12
    Geda YE, Roberts RO, Mielke MM, et al. Baseline neuropsychiatric symptoms and the risk of incident mild cognitive impairment: a population-based study. Am J Psychiatry 2014;171(05):572-581
  • 13
    Ismail Z, Smith EE, Geda Y, et al; ISTAART Neuropsychiatric Symptoms Professional Interest Area. Neuropsychiatric symptoms as early manifestations of emergent dementia: Provisional diagnostic criteria for mild behavioral impairment. Alzheimers Dement 2016;12(02):195-202
  • 14
    Taragano FE, Allegri RF, Lyketsos C. Mild behavioral impairment: A prodromal stage of dementia. Dement Neuropsychol 2008;2(04):256-260
  • 15
    Taragano FE, Allegri RF, Krupitzki H, et al. Mild behavioral impairment and risk of dementia: a prospective cohort study of 358 patients. J Clin Psychiatry 2009;70(04):584-592
  • 16
    Taragano FE, Allegri RF, Heisecke SL, et al. Risk of Conversion to Dementia in a Mild Behavioral Impairment Group Compared to a Psychiatric Group and to a Mild Cognitive Impairment Group. J Alzheimers Dis 2018;62(01):227-238
  • 17
    Ismail Z, Agüera-Ortiz L, Brodaty H, et al; NPS Professional Interest Area of the International Society of to Advance Alzheimer’s Research and Treatment (NPS-PIA of ISTAART) The Mild Behavioral Impairment Checklist (MBI-C): A Rating Scale for Neuropsychiatric Symptoms in Pre-Dementia Populations. J Alzheimers Dis 2017;56(03):929-938
  • 18
    Sperling RA, Aisen PS, Beckett LA, et al. Toward defining the preclinical stages of Alzheimer’s disease: recommendations from the National Institute on Aging-Alzheimer’s Association workgroups on diagnostic guidelines for Alzheimer’s disease. Alzheimers Dement 2011;7(03):280-292
  • 19
    Reisberg B, Monteiro I, Torossian C, et al. The BEHAVE-AD assessment system: a perspective, a commentary on new findings, and a historical review. Dement Geriatr Cogn Disord 2014;38;(1-2):89-146
  • 20
    Alexopoulos GS, Abrams RC, Young RC, Shamoian CA. Cornell Scale for Depression in Dementia. Biol Psychiatry 1988;23(03):271-284
  • 21
    Mathys M. Pharmacologic management of behavioral and psychological symptoms of major neurocognitive disorder. Ment Health Clin 2018;8(06):284-293
  • 22
    Kales HC, Gitlin LN, Lyketsos CG. Assessment and management of behavioral and psychological symptoms of dementia. BMJ 2015;350:h369
  • 23
    Canevelli M, Adali N, Voisin T, et al. Behavioral and psychological subsyndromes in Alzheimer’s disease using the Neuropsychiatric Inventory. Int J Geriatr Psychiatry 2013;28(08):795-803
  • 24
    Kales HC, Lyketsos CG, Miller EM, Ballard C. Management of behavioral and psychological symptoms in people with Alzheimer’s disease: an international Delphi consensus. Int Psychogeriatr 2019;31(01):83-90
  • 25
    Abraha I, Rimland JM, Trotta FM, et al. Systematic review of systematic reviews of non-pharmacological interventions to treat behavioural disturbances in older patients with dementia. The SENATOR-OnTop series. BMJ Open 2017;7(03):e012759
  • 26
    Reus VI, Fochtmann LJ, Eyler AE, et al. The American Psychiatric Association Practice Guideline on the Use of Antipsychotics to Treat Agitation or Psychosis in Patients With Dementia. Am J Psychiatry 2016;173(05):543-546
  • 27
    Borsje P, Wetzels RB, Lucassen PL, Pot AM, Koopmans RT. The course of neuropsychiatric symptoms in community-dwelling patients with dementia: a systematic review. Int Psychogeriatr 2015;27(03):385-405
  • 28
    van der Linde RM, Dening T, Stephan BC, Prina AM, Evans E, Brayne C. Longitudinal course of behavioural and psychological symptoms of dementia: systematic review. Br J Psychiatry 2016; 209(05):366-377
  • 29
    Tampi RR, Jeste DV. Dementia Is More Than Memory Loss: Neuropsychiatric Symptoms of Dementia and Their Nonpharmacological and Pharmacological Management. Am J Psychiatry 2022;179(08):528-543
  • 30
    Caramelli P, Marinho V, Laks J, et al. Treatment of dementia: recommendations of the Scientific Department of Cognitive Neurology and Aging of the Brazilian Academy of Neurology. Dement Neuropsychol 2022;16(03, Suppl 1):88-100
  • 31
    Tan ECK, Johnell K, Bell JS, et al. Do Acetylcholinesterase Inhibitors Prevent or Delay Psychotropic Prescribing in People With Dementia? Analyses of the Swedish Dementia Registry. Am J Geriatr Psychiatry 2020;28(01):108-117
  • 32
    Xu H, Garcia-Ptacek S, Jönsson L, Wimo A, Nordström P, Eriksdotter M. Long-term Effects of Cholinesterase Inhibitors on Cognitive Decline and Mortality. Neurology 2021;96(17):e2220-e2230
  • 33
    Marucci G, Buccioni M, Ben DD, Lambertucci C, Volpini R, Amenta F. Efficacy of acetylcholinesterase inhibitors in Alzheimer’s disease. Neuropharmacology 2021;190:108352
  • 34
    Teixeira AL, Gonzales MM, de Souza LC, Weisenbach SL. Revisiting Apathy in Alzheimer’s Disease: From Conceptualization to Therapeutic Approaches. Behav Neurol 2021;2021:6319826
  • 35
    Salem H, Suchting R, Gonzales MM, Seshadri S, Teixeira AL. Apathy as a Predictor of Conversion from Mild Cognitive Impairment to Alzheimer’s Disease: A Texas Alzheimer’s Research and Care Consortium (TARCC) Cohort-Based Analysis. J Alzheimers Dis 2023;92(01):129-139
  • 36
    Miller DS, Robert P, Ereshefsky L, et al. Diagnostic criteria for apathy in neurocognitive disorders. Alzheimers Dement 2021;17(12):1892-1904
  • 37
    Husain M, Roiser JP. Neuroscience of apathy and anhedonia: a transdiagnostic approach. Nat Rev Neurosci 2018;19(08):470-484
  • 38
    Sassi KLM, Rocha NP, Colpo GD, John V, Teixeira AL. Amphetamine Use in the Elderly: A Systematic Review of the Literature. Curr Neuropharmacol 2020;18(02):126-135
  • 39
    Mintzer J, Lanctôt KL, Scherer RW, et al; ADMET 2 Research Group. Effect of Methylphenidate on Apathy in Patients With Alzheimer Disease: The ADMET 2 Randomized Clinical Trial. JAMA Neurol 2021;78(11):1324-1332
  • 40
    Burke AD, Goldfarb D, Bollam P, Khokher S. Diagnosing and Treating Depression in Patients with Alzheimer’s Disease. Neurol Ther 2019;8(02):325-350
  • 41
    Asmer MS, Kirkham J, Newton H, et al. Meta-Analysis of the Prevalence of Major Depressive Disorder Among Older Adults With Dementia. J Clin Psychiatry 2018;79(05):17r11772
  • 42
    Olin JT, Schneider LS, Katz IR, et al. Provisional diagnostic criteria for depression of Alzheimer disease. Am J Geriatr Psychiatry 2002;10(02):125-128
  • 43
    Agüera-Ortiz L, García-Ramos R, Grandas Pérez FJ, et al. Depression in Alzheimer’s Disease: A Delphi Consensus on Etiology, Risk Factors, and Clinical Management. Front Psychiatry 2021;12:638651
  • 44
    Mendez MF. The Relationship Between Anxiety and Alzheimer’s Disease. J Alzheimers Dis Rep 2021;5(01):171-177
  • 45
    Cummings J, Pinto LC, Cruz M, et al. Criteria for Psychosis in Major and Mild Neurocognitive Disorders: International Psychogeriatric Association (IPA) Consensus Clinical and Research Definition. Am J Geriatr Psychiatry 2020;28(12):1256-1269
  • 46
    Sano M, Cummings J, Jeste DV, Finkel S, Reichman W. International Psychogeriatric Association (IPA) consensus for defining psychosis in major and mild neurocognitive disorders. Int Psychogeriatr 2022;34(03):203-207
  • 47
    Ropacki SA, Jeste DV. Epidemiology of and risk factors for psychosis of Alzheimer’s disease: a review of 55 studies published from 1990 to 2003. Am J Psychiatry 2005;162(11):2022-2030
  • 48
    Pearce D, Gould RL, Roughley M, et al. Paranoid and misidentification subtypes of psychosis in dementia. Neurosci Biobehav Rev 2022;134:104529
  • 49
    Nicolato R, Corrêa H, Romano-Silva MA, Teixeira AL Jr. Delusional parasitosis or Ekbom syndrome: a case series. Gen Hosp Psychiatry 2006;28(01):85-87
  • 50
    Wang PS, Schneeweiss S, Avorn J, et al. Risk of death in elderly users of conventional vs. atypical antipsychotic medications. N Engl J Med 2005;353(22):2335-2341
  • 51
    Yury CA, Fisher JE. Meta-analysis of the effectiveness of atypical antipsychotics for the treatment of behavioural problems in persons with dementia. Psychother Psychosom 2007;76(04):213-218
  • 52
    Schneider LS, Tariot PN, Dagerman KS, et al; CATIE-AD Study Group. Effectiveness of atypical antipsychotic drugs in patients with Alzheimer’s disease. N Engl J Med 2006;355(15):1525-1538
  • 53
    Vigen CL, Mack WJ, Keefe RS, et al. Cognitive effects of atypical antipsychotic medications in patients with Alzheimer’s disease: outcomes from CATIE-AD. Am J Psychiatry 2011;168(08):831-839
  • 54
    Maher AR, Maglione M, Bagley S, et al. Efficacy and comparative effectiveness of atypical antipsychotic medications for off-label uses in adults: a systematic review and meta-analysis. JAMA 2011;306(12):1359-1369
  • 55
    Yunusa I, Alsumali A, Garba AE, Regestein QR, Eguale T. Assessment of Reported Comparative Effectiveness and Safety of Atypical Antipsychotics in the Treatment of Behavioral and Psychological Symptoms of Dementia: A Network Meta-analysis. JAMA Netw Open 2019;2(03):e190828
  • 56
    Yunusa I, Rashid N, Demos GN, Mahadik BS, Abler VC, Rajagopalan K. Comparative Outcomes of Commonly Used Off-Label Atypical Antipsychotics in the Treatment of Dementia-Related Psychosis: A Network Meta-analysis. Adv Ther 2022;39(05): 1993-2008
  • 57
    Kirkham J, Sherman C, Velkers C, et al. Antipsychotic Use in Dementia. Can J Psychiatry 2017;62(03):170-181
  • 58
    Cummings J, Isaacson S, Mills R, et al. Pimavanserin for patients with Parkinson’s disease psychosis: a randomised, placebo-controlled phase 3 trial. Lancet 2014;383(9916):533-540
  • 59
    Tariot PN, Cummings JL, Soto-Martin ME, et al. Trial of Pimavanserin in Dementia-Related Psychosis. N Engl J Med 2021;385(04):309-319
  • 60
    Moreno GM, Gandhi R, Lessig SL, Wright B, Litvan I, Nahab FB. Mortality in patients with Parkinson disease psychosis receiving pimavanserin and quetiapine. Neurology 2018;91(17):797-799
  • 61
    Ismail Z, Creese B, Aarsland D, et al. Psychosis in Alzheimer disease - mechanisms, genetics and therapeutic opportunities. Nat Rev Neurol 2022;18(03):131-144
  • 62
    Sano M, Cummings J, Auer S, et al. Agitation in cognitive disorders: Progress in the International Psychogeriatric Association consensus clinical and research definition. Int Psychogeriatr 2023; •••:1-13
  • 63
    Cummings J, Sano M, Auer S, et al. Reduction and prevention of agitation in persons with neurocognitive disorders: an international psychogeriatric association consensus algorithm. Int Psychogeriatr 2023;•••:1-12
  • 64
    Sano M, Zhu CW, Neugroschl J, Grossman HT, Schimming C, Aloysi A. Agitation in Cognitive Disorders: Use of the National Alzheimer’s Coordinating Center Uniform Data Set (NACC-UDS) to Evaluate International Psychogeriatric Association Definition. Am J Geriatr Psychiatry 2022;30(11):1198-1208
  • 65
    Selbaek G, Engedal K, Benth JŠ, Bergh S. The course of neuropsychiatric symptoms in nursing-home patients with dementia over a 53-month follow-up period. Int Psychogeriatr 2014;26(01):81-91
  • 66
    Livingston G, Kelly L, Lewis-Holmes E, et al. Non-pharmacological interventions for agitation in dementia: systematic review of randomised controlled trials. Br J Psychiatry 2014;205(06):436-442
  • 67
    Watt JA, Goodarzi Z, Veroniki AA, et al. Comparative Efficacy of Interventions for Aggressive and Agitated Behaviors in Dementia: A Systematic Review and Network Meta-analysis. Ann Intern Med 2019;171(09):633-642
  • 68
    Salem H, Nagpal C, Pigott T, Teixeira AL. Revisiting Antipsychotic-induced Akathisia: Current Issues and Prospective Challenges. Curr Neuropharmacol 2017;15(05):789-798
  • 69
    Banerjee S, High J, Stirling S, et al. Study of mirtazapine for agitated behaviours in dementia (SYMBAD): a randomised, double-blind, placebo-controlled trial. Lancet 2021;398(10310):1487-1497
  • 70
    Porsteinsson AP, Drye LT, Pollock BG, et al; CitAD Research Group. Effect of citalopram on agitation in Alzheimer disease: the CitAD randomized clinical trial. JAMA 2014;311(07):682-691
  • 71
    Schneider LS, Frangakis C, Drye LT, et al; CitAD Research Group. Heterogeneity of Treatment Response to Citalopram for Patients With Alzheimer’s Disease With Aggression or Agitation: The CitAD Randomized Clinical Trial. Am J Psychiatry 2016;173(05):465-472
  • 72
    Administration, F.a.D. FDA Approves First Drug to Treat Agitation Symptoms Associated with Dementia due to Alzheimer's Disease 2023 05/11/2023 [cited 2023.
  • 73
    Davies SJ, Burhan AM, Kim D, et al. Sequential drug treatment algorithm for agitation and aggression in Alzheimer's and mixed dementia. J Psychopharmacol 2018;32(05):509-523
  • 74
    Supasitthumrong T, Bolea-Alamanac BM, Asmer S, Woo VL, Abdool PS, Davies SJC. Gabapentin and pregabalin to treat aggres- sivity in dementia: a systematic review and illustrative case report. Br J Clin Pharmacol 2019;85(04):690-703
  • 75
    Cummings JL, Lyketsos CG, Peskind ER, et al. Effect of Dextrome-thorphan-Quinidine on Agitation in Patients With Alzheimer Disease Dementia: A Randomized Clinical Trial. JAMA 2015;314(12):1242-1254
  • 76
    Herrmann N, Ruthirakuhan M, Gallagher D, et al. Randomized Placebo-Controlled Trial of Nabilone for Agitation in Alzheimer’s Disease. Am J Geriatr Psychiatry 2019;27(11):1161-1173
  • 77
    Ruthirakuhan M, Lanctôt KL, Vieira D, Herrmann N. Natural and Synthetic Cannabinoids for Agitation and Aggression in Alzheimer’s Disease: A Meta-Analysis. J Clin Psychiatry 2019;80(02):18r12617
  • 78
    Herrmann N, Wang HJ, Song BX, Bawa KK, Lanctôt KL. Risks and benefits of current and novel drugs to treat agitation in Alzheimer’s disease. Expert Opin Drug Saf 2022;21(10):1289-1301
  • 79
    Shi L, Chen SJ, Ma MY, et al. Sleep disturbances increase the risk of dementia: A systematic review and meta-analysis. Sleep Med Rev 2018;40:4-16
  • 80
    Bubu OM, Brannick M, Mortimer J, et al. Sleep, Cognitive impairment, and Alzheimer’s disease: A Systematic Review and Meta-Analysis. Sleep 2017;40(01):
  • 81
    Casagrande M, Forte G, Favieri F, Corbo I. Sleep Quality and Aging: A Systematic Review on Healthy Older People, Mild Cognitive Impairment and Alzheimer’s Disease. Int J Environ Res Public Health 2022;19(14):8457
  • 82
    Javed B, Javed A, Kow CS, Hasan SS. Pharmacological and non-pharmacological treatment options for sleep disturbances in Alzheimer’s disease. Expert Rev Neurother 2023;23(06):501-514
  • 83
    Wade AG, Farmer M, Harari G, et al. Add-on prolonged-release melatonin for cognitive function and sleep in mild to moderate Alzheimer’s disease: a 6-month, randomized, placebo-controlled, multicenter trial. Clin Interv Aging 2014;9:947-961
  • 84
    Serfaty M, Kennell-Webb S, Warner J, Blizard R, Raven P. Double blind randomised placebo controlled trial of low dose melatonin for sleep disorders in dementia. Int J Geriatr Psychiatry 2002;17 (12):1120-1127
  • 85
    Gehrman PR, Connor DJ, Martin JL, Shochat T, Corey-Bloom J, Ancoli-Israel S. Melatonin fails to improve sleep or agitation in double-blind randomized placebo-controlled trial of institutionalized patients with Alzheimer disease. Am J Geriatr Psychiatry 2009;17(02):166-169
  • 86
    McCleery J, Sharpley AL. Pharmacotherapies for sleep disturbances in dementia. Cochrane Database Syst Rev 2020;11(11):CD009178

Publication Dates

  • Publication in this collection
    15 Jan 2024
  • Date of issue
    Dec 2023

History

  • Received
    15 Sept 2023
  • Reviewed
    05 Nov 2023
  • Accepted
    08 Nov 2023
Academia Brasileira de Neurologia - ABNEURO R. Vergueiro, 1353 sl.1404 - Ed. Top Towers Offices Torre Norte, 04101-000 São Paulo SP Brazil, Tel.: +55 11 5084-9463 | +55 11 5083-3876 - São Paulo - SP - Brazil
E-mail: revista.arquivos@abneuro.org